You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/11282024

The Immunobiology of Schistosomiasis

Article  in  Nature reviews. Immunology · August 2002


DOI: 10.1038/nri843 · Source: PubMed

CITATIONS READS
745 926

2 authors, including:

Andrew S MacDonald
The University of Manchester
118 PUBLICATIONS   4,561 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Podosome biology View project

All content following this page was uploaded by Andrew S MacDonald on 19 May 2014.

The user has requested enhancement of the downloaded file.


REVIEWS

THE IMMUNOBIOLOGY OF
SCHISTOSOMIASIS
Edward J. Pearce and Andrew S. MacDonald
Schistosomes are parasitic worms that are a prime example of a complex multicellular
pathogen that flourishes in the human host despite the development of a pronounced immune
response. Understanding how the immune system deals with such pathogens is a daunting
challenge. The past decade has seen the use of a wide range of new approaches to determine
the nature and function of the immune response to schistosomes. Here, we attempt to
summarize advances in our understanding of the immunology of schistosomiasis, with the bulk
of the review reflecting the experimental focus on Schistosoma mansoni infection in mice.

DIGENETIC TREMATODE Schistosoma is a genus of parasitic DIGENETIC TREMATODES and co-infection status all influence the development of
Digenetic trematodes, or flukes, that chronically infects more than 200 million people in the immune response and, so, disease severity.
are extremely successful developing countries (BOX 1). The estimated mortality Two main clinical conditions are recognized in
parasitic worms, the life cycle of
which requires development in
owing to Schistosoma mansoni and Schistosoma haema- S. mansoni-infected individuals — acute schistosomiasis
at least two hosts. Importantly, tobium in sub-Saharan Africa is 280,000 per year1. and chronic schistosomiasis.
they can parasitize all classes of Three early findings piqued the interest of immunolo-
vertebrate, causing widespread gists in schistosomiasis: the immune response is inti- Acute schistosomiasis: a TH1 disease? Acute schistosomi-
medical and economic
mately involved in the development of many of the asis in humans is a debilitating febrile illness (Katayama
problems.
pathological changes that accompany infection; fever) that can occur before the appearance of eggs in
T HELPER 1/T HELPER 2 infected individuals can have resistance to superinfec- the stool and which is thought generally to peak
(TH1/TH2). Subsets of CD4+ tion; and schistosomes survive for years in the host between 6 and 8 weeks after infection4. During acute ill-
T cells that are characterized by despite a strong immune response. More recently, inter- ness, which is less well studied than chronic disease (see
their cytokine-production
profiles. TH1 cells primarily
est in these parasites has increased owing to demonstra- below), there is a measurable level of tumour-necrosis
produce IFN-γ, and generally tions that schistosome maturation and fecundity are, in factor (TNF) in the plasma, and peripheral-blood
provide protection against some way, dependent on the host immune response. mononuclear cells (PBMCs) produce large quantities of
intracellular pathogens, whereas Schistosomes, like other parasitic helminths, induce TNF, interleukin-1 (IL-1) and IL-6 (REF. 5). Notably,
TH2 cells mainly produce IL-4,
marked T HELPER 2 (TH2) responses, providing a model cytokine production by PBMCs after stimulation with
IL-5 and IL-13, and are
important for immunity to system for studying the development and function of parasite antigen reflects a dominant T HELPER 1 (TH1),
helminth parasites. this type of immune response. rather than TH2, response5. Presumably, in the natural
progression of the disease, the developing egg-antigen-
Immune-related pathologies during infection induced TH2 response downregulates the production
Schistosomiasis causes a range of morbidities, the and effector functions of these pro-inflammatory medi-
Department of Pathobiology, development of which seems to be influenced to a large ators (FIG. 1); the production of IL-10 during this period
University of Pennsylvania, extent by the nature of the induced immune response might have a crucial role in this process6.
Philadelphia, Philadelphia and its effects on granuloma formation and associated Anomalously, the febrile illness that is associated
19104-6008, USA. pathologies in target organs2,3 (FIG. 1; BOX 2). Field stud- with the initial stages of schistosome infection seems to
Correspondence to E.J.P.
e-mail: ejpearce@mail.med.
ies in endemic areas, combined with animal experi- be uncommon in individuals who live in areas that are
upenn.edu ments, have led to the view that host genetics, infection endemic for schistosomiasis. It occurs, instead, in indi-
doi:10.1038/nri843 intensity, in utero sensitization to schistosome antigen viduals who have no previous history of exposure who

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 4 9 9


REVIEWS

Box 1 | The biology of schistosomes


Of the ~2700 genera of Digenian parasites, the 13 that comprise the Schistosomatidae are unusual in four ways: they
have two rather than three hosts; they are dioecious (having male and female reproductive organs in separate
individuals), rather than hermaphrodite or asexual; they infect their hosts by directly penetrating the body surface,
rather than as a result of being eaten; and they parasitize the intravascular niche138. As for all sexual Digeneans, there is
an alternation of generations, such that asexual reproduction occurs in the intermediate (snail) host and sexual
reproduction occurs in the definitive (mammalian) host. The life cycle of Schistosoma mansoni is shown in the figure.
S. mansoni lives for long periods, with no evidence of immune-mediated clearance of adult worms139, and it has
evolved to use host factors for developmental signalling. By contrast, there is evidence for the immune-mediated
killing of adult Schistosoma haematobium parasites over time139. Infection is initiated by cercariae, which burrow into
the skin, transform into schistosomula, and then enter the vasculature and migrate to the portal system, where they
mature into adult worms. Eggs, which have tough shells, are released by female parasites within the vasculature; they
cross the endothelium and basement membrane of the vein, and traverse the intervening tissue, basement membrane
and epithelium of the intestine (S. mansoni and Schistosoma japonicum) or bladder (S. haematobium) en route to the
exterior. It is not clear yet how this process occurs, although there seems to be an immunological component, because
egg excretion is minimal in immunocompromised mice, but can be increased by the transfer of sera or lymphocytes
from infected animals140. Moreover, in a comparison of S. mansoni-infected HIV+ and HIV− patients, a correlation
between diminished egg excretion and decreased CD4+ T-cell counts was apparent141. It is unclear how eggs initially
attach to the endothelium and initiate penetration during extravasation, although factors that are released from
platelets in response to the eggs seem to be involved140,142.
So far, it has proved to be impossible to culture schistosomes through their complete life cycle in vitro, and there are no
published reports of techniques for routinely expressing transgenes in schistosomes or for targeted gene silencing. Also,
there are no schistosome cell lines. So, analyses of schistosome–host interactions rely on host-focused interventions and
traditional parasitological techniques.

Host-derived mediators
(TGF-β, IL-7, TNF ?)

CD4+
? T cells

Receptors for host-


derived mediators

Liver Adult female Adult male

Portal vein

Intestine

Fresh water
Eggs
(~140 µm)

Cercariae
(~800 µm)

Miracidia
Snail (~180 µm)
Intermediate host

IL-7, interleukin-7; TGF-β, transforming growth factor-β; TNF, tumour-necrosis factor.

500 | JULY 2002 | VOLUME 2 www.nature.com/reviews/immunol


REVIEWS

Intensity of immune response


TH1 TH 2

Weeks after 3 6 8 12
infection
Acute Chronic

Schistosomula Eggs
Cercariae
Adult
worms
Figure 1 | Development of the immune response in infection. In the course of an infection, the immune response
progresses through at least three phases. In the first 3–5 weeks, during which the host is exposed to migrating immature
parasites, the dominant response is T helper 1 (TH1)-like. As the parasites mature, mate and begin to produce eggs at weeks
5–6, the response alters markedly; the TH1 component decreases and this is associated with the emergence of a strong TH2
response. This response is induced primarily by egg antigens. During the chronic phase of infection (infections are long lived
and worms continue to produce eggs — ~300 per day in the case of each Schistosoma mansoni female), the TH2 response is
modulated and granulomas that form around newly deposited eggs are smaller than at earlier times during infection. From
work in the mouse, there now seems to be a correlation between the inability to form granulomas, or the development and
persistence of a highly pro-inflammatory TH1-like response beyond the acute phase, and the development of hepatotoxic liver
disease150. By contrast, TH2-cell-mediated granulomas seem to protect hepatocytes, but allow the development of fibrosis3,150.
Although it is clear that severe fibrosis occurs in human schistosomiasis, there is debate over the existence of the hepatotoxic
form of disease3,150. TH2 responses are also strongly implicated in naturally acquired resistance to reinfection with
schistosomes.

become infected after travelling into an endemic area. but pathological changes in the intestine were more evi-
One explanation for this difference is that individuals dent in the absence of IL-4 — non-haemorrhagic lesions
can become sensitized to schistosomes in utero as a on the mucosal surface9 were associated with the ineffi-
result of maternal infection, which subsequently allows cient passage of eggs into the lumen10. This process was
them to respond differently from ‘naive’ individuals accompanied by detectable levels of lipopolysaccharide
when they themselves become infected. Data from in the plasma, perhaps owing to the translocation of
analyses of cord-blood lymphocytes taken from the intestinal bacteria10. Analyses of the immune responses
babies of infected and uninfected mothers support the of infected Il4 –/– mice showed that there was a correla-
view that in utero sensitization does occur and, more- tion between elevated levels of nitric oxide (NO) and
over, indicate that the fetal response is phenotypically disease severity9. Treatment with uric acid, which is a
similar to the response of the mother7,8. The pre-existing peroxyradical scavenger, had marked ameliorative
TH2 response in such children might make them less effects11, which indicates that a combination of reactive
likely to develop a pro-inflammatory response on first oxygen and nitrogen intermediates might have a role in
infection with schistosomes. acute disease.
An examination of disease in mice has shown that an
inability to develop a TH2 response to regulate the initial Chronic schistosomiasis: a TH2 disease? Chronic disease is
pro-inflammatory response that is associated with acute graded according to severity. The most serious form is a
schistosomiasis is lethal. This first became apparent life-threatening hepatosplenic disease, which is usually
when C57BL/6 Il4 −/− mice were infected with S. mansoni. accompanied by severe hepatic and periportal fibrosis,
Coincident with the onset of parasite egg production in portal hypertension and portosystemic shunting of
these animals, a condition that was similar to severe acute venous blood2 (BOX 2). Fibrosis itself is ranked on the basis
schistosomiasis in humans developed, which was charac- of ultrasound patterns that provide a quantitative tool for
terized by cachexia and significant mortality9,10. These assessing the severity of disease12.
mice developed relatively normal hepatic granulomas Although TH2 responses seem to have a crucial role in
(although they lacked an eosinophil component; BOX 2), modulating potentially life-threatening disease during

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 5 0 1


REVIEWS

Box 2 | The granuloma


In infection with any schistosome species, chronic disease is the result of the ongoing host response to accumulating
tissue-trapped eggs. In Schistosoma mansoni and Schistosoma japonicum infections, the liver is the principal site that is
affected, because many of the eggs are carried by the blood flow into this organ, the sinusoids of which are too small for
the eggs to traverse. This is a dead-end for the eggs, which eventually die within the tissue. Intestinal damage by
traversing eggs can also be problematic. During Schistosoma haematobium infection, the passage of eggs across the
bladder wall causes damage to this organ. The CD4+ T-cell response that is induced by egg antigens orchestrates the
development of granulomatous lesions — which are composed of collagen fibres and cells, including macrophages,
eosinophils and CD4+ T cells — around the individual eggs2,48 (see figure). As the eggs die, the granulomas resolve,
leaving fibrotic plaques. Severe consequences of infection with S. mansoni and S. japonicum are the result of an increase
in portal blood pressure as the liver becomes fibrotic, congested and harder to perfuse. Under these conditions, the
diameter of the portal vein increases and the wall of the portal vein becomes fibrotic. Associated with these changes is
the development of ascites (the accumulation of serous
Hepatocytes
fluid in the peritoneal cavity) and portal–systemic
venous shunts (new blood vessels that bypass the liver),
which can rupture, leading to life-threatening bleeding.
The most serious effects of infection with
S. haematobium are bladder cancer143 and genital
schistosomiasis, a condition in which eggs pass through
the cervix in women or into the testes in men144,145.
Paradoxically, granulomas might have an essential
host-protective role. In mice that were tolerized against
S. mansoni egg antigen, granuloma development did not
Schistosome
occur during infection and the animals had severe egg
hepatotoxic liver damage, which was evident as
microvesicular lipid accumulations (or steatoses)
within hepatocytes146. This is thought to be mediated
by hepatotoxins that are secreted from eggs, and the
granuloma, together with egg-antigen-specific
antibodies (which might act in a neutralizing capacity),
is envisaged as sequestering these toxins away from
hepatocytes147. A central role for tumour-necrosis factor
(TNF) in the development of the granuloma has been
Collagen CD4+ T cell
proposed on the basis of one finding that the injection
of TNF into infected severe combined immunodeficient Eosinophil Other cell
(SCID) mice is sufficient to allow the development of a Macrophage
focal lesion around parasite eggs43 (but, see REF. 47).

the initial stages of schistosomiasis, prolonged TH2 endemic, Dessein and colleagues found that severe
responses contribute to the development of hepatic hepatic fibrosis (as identified by ultrasound) was more
fibrosis and chronic morbidity3. The main TH2 cytokine likely to occur in certain families20. A SEGREGATION ANALYSIS
that is responsible for fibrosis is IL-13. So, schistosome- showed that a codominant major gene, known as SM2,
infected mice in which IL-13 is either absent (Il13 −/−)10, is responsible for the observed familial distribution of
ineffective (IL-4 receptor α-chain-knockouts; Il4rα −/−)13 hepatic fibrosis and portal hypertension.‘Informative’
or neutralized by treatment with soluble IL-13Rα2–Fc14 families, which had multiple cases of severe fibrosis,
fail to develop the severe hepatic fibrosis that normally were used to map SM2 to 6q22–q23 — a region that
occurs during infection, which leads to prolonged sur- contains the gene that encodes IFN-γ receptor 1 (IFN-
vival of these mice10. The mechanism by which IL-13 is γR1)20. One interpretation of these data is that muta-
able to promote fibrogenesis has been elucidated in a tions in IFN-γR1 that lead to loss of function of the
series of recent studies15–18 (FIG. 2). These findings might receptor are associated with a lack of effectiveness of
have implications beyond schistosomiasis for the possi- IFN-γ in suppressing fibrogenesis.
SEGREGATION ANALYSIS ble use of IL-13-blocking therapies in other fibrotic dis- It is not clear yet whether IL-13 is important for
This technique is used to predict eases. Mediators that are associated with TH1 responses, hepatic fibrosis in human schistosomiasis. Most humans
the probability that certain
such as interferon-γ (IFN-γ), IL-12, TNF and NO can who are infected with schistosomes develop TH2
individuals will be of a certain
genotype given information prevent IL-13-mediated fibrosis18 (FIG. 2). responses21,22, but, as expected in an outbred population,
about the genotypes of ancestors Infection intensity is one factor that can affect the the intensity of the response differs between individuals.
and assumptions about the severity of chronic schistosomatic disease, perhaps par- On the basis of the amount of IFN-γ or IL-5 (or other
mode of inheritance. It can be ticularly in children (see below)19. However, it seems to TH2 cytokines) that is produced by PBMCs in response
used to distinguish between
different models of inheritance
be more important whether an infected individual is to antigen, some individuals do seem to have a more
(for example, major gene versus genetically predisposed to disease19,20. In an analysis of TH1-like response. In one of the few studies that have
multifactorial). pedigrees in Sudan, in an area where S. mansoni is attempted systematically to correlate the immune

502 | JULY 2002 | VOLUME 2 www.nature.com/reviews/immunol


REVIEWS

Although it is thought generally that, during schisto-


IL-12
somiasis, immunopathology and immunoregulation
are under the control of egg-antigen-specific TH cells,
there is a large body of data that indicates the impor-
IFN-γ TNF tance of IDIOTYPIC REGULATION in these processes (this
Macrophage complex area is reviewed succinctly in a recent paper by
Nitric oxide the only group that continues to work in this area25)
+ (BOX 3). So, egg-specific antibodies (idiotypes) that are

L- hydroxy- Citrulline purified from the sera of patients with less severe
arginine chronic disease are able to stimulate T cells from the
Inducible same patients to proliferate. Rabbit antisera that are
nitric oxide
synthase raised against these idiotypes bind well to idiotypes
L-arginine from chronically infected mice and, in so doing, define
crossreactive idotypes (CRIs) that cannot be detected in
the sera of patients with hepatosplenomegaly or in an
Arginase
Collagen unusual subset of infected male CBA/J mice that (for
L-ornithine Proline synthesis
and fibrosis reasons that are not clear) develop a condition that is
Ornithine–
+ aminotransferase analogous to hepatosplenic disease26. Moreover, the
early appearance of CRIs in infected male CBA/J mice is
a robust predictor that the animals will not develop
severe disease, whereas infected male CBA/J mice that
IL-4 fail to develop CRIs invariably die early after the onset of
egg production or develop severe chronic disease. In
IL-13 addition, neonatal mice that are injected with CRIs
develop a regulatory anti-idiotypic T-cell response that,
Figure 2 | IL-13 and IFN-γ/IL-12 counter-regulate macrophage-activation status and in later life, has a role in preventing the development of
control fibrosis. The fibrogenic role of interleukin-13 (IL-13) seems to stem from its ability, severe morbidity during chronic infection27. In keeping
together with IL-4, to induce the expression of arginase in macrophages18. Arginase uses with the data that are outlined above — which indicate
L-arginine as a substrate to make L-ornithine, which is converted to proline by ornithine-
that IFN-γ might have a host-protective role in the mit-
aminotransferase. Proline is an essential amino acid that is involved in collagen production and,
therefore, in the development of fibrosis. Fibrosis is inhibited in mice that are immunized with egg
igation of severe chronic disease — the neonatal mice
antigens plus IL-12; cytokines that are produced as components of the induced TH1 response that were injected with CRIs developed a population of
(such as IFN-γ and TNF) prevent TH2-response development (and, so, IL-13 production) and also T cells that were able to produce IFN-γ in response to
activate macrophages to express inducible nitric oxide synthase (iNOS), rather than arginase. schistosome egg antigen. Findings from these studies of
This immunization protocol is ineffective in iNOS-knockout mice, despite the induction of excellent idiotypic regulatory pathways are remarkably consistent
TH1 responses in these animals. This seems to be due to the fact that iNOS uses arginine to between human and mouse schistosomiasis25.
make nitric oxide (NO) and citrulline — an intermediate in this pathway is L-hydroxyarginine, which
inhibits arginase, effectively reducing the amount of proline that is available for collagen synthesis.
These findings fit nicely with early work in this area, which showed that IFN-γ could have an anti- The importance of a balanced TH response. Interestingly,
fibrogenic role in the schistosome granuloma, as well as in other conditions. Adapted from REF. 18. the severe disease that is observed in infected Il4 −/− mice
IFN-γ, interferon-γ; TNF, tumour-necrosis factor. (see above) is not related to increased parasite burden,
but, rather, seems to be linked to the immunological
consequences of the absence of TH2 cytokines. So, an
response with disease severity, patients with hepato- important function of the TH2 response during infec-
splenomegaly owing to S. mansoni infection were found tion is to produce cytokines that can prevent or dampen
to have a TH1-like response and high plasma levels of the production or effector functions of potentially dan-
TNF receptor I (TNFRI) and TNFRII, whereas individ- gerous inflammatory mediators. A more comprehen-
uals who had less severe disease but similarly intense sive view of this issue has emerged recently from a
infections (as assessed by counting the number of eggs detailed investigation of the outcome of infection in
in faecal samples) had TH2 responses and low plasma IL-4 and IL-10, IL-12 and IL-10, and IFN-γ and IL-10
levels of soluble TNFR23. The finding that severe chronic double-deficient mice 28,29. IL-10 has been considered
disease is associated with TH1, rather than TH2, responses primarily to be a regulator of pro-inflammatory
contrasts with data from the mouse model. However, responses, and on the basis of the findings in the Il4 −/−
IDIOTYPIC REGULATION hepatosplenic fibrosis in the patients that were used in mice, it was thought that its absence would result in
The antigen-binding site of an this study was not assessed by ultrasound, and it is now increased disease severity during schistosomiasis.
antibody is an idiotype. As an
clear that hepatosplenomegaly is not always accompa- Consistent with this, during infection, the Il4 −/−Il10 −/−
immune response develops and
clonal B-cell expansion occurs, nied by severe fibrosis24. So, clarification of the role of animals developed highly polarized TH1 responses and
the prevalence of this previously TH1 responses in severe chronic human schistosomiasis a lethal acute wasting condition that seemed to be an
rare idiotype increases and can will require detailed analyses of immune responses in exaggerated form of the disease that is observed in
lead to the development of an patients who have been carefully assessed by ultrasound. infected Il4 −/− animals (see above), with evidence of
anti-idiotypic T- and B-cell
response. Focus on this once
Taking into account this caveat, genetic and immunolog- increased hepatoxicity28. Perhaps surprisingly, IL-12
popular area of immunology is ical studies in mice and humans do indicate that IFN-γ is and IL-10, and IFN-γ and IL-10 double-deficient mice,
now minimal. important for fibrosis during schistosomiasis. in contrast to Il12 −/− or Ifnγ −/− mice, also developed

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 5 0 3


REVIEWS

a strong TH1 response and were sensitized to develop a


Box 3 | Some important understudied and/or unresolved areas
lethal acute disease with severe hepatotoxicity on subse-
• The mechanism that underlies idiotypic regulation in schistosomiasis. quent infection34. One possible explanation is that IL-12,
• The role of regulatory T cells148 during schistosomiasis. The magnitude of T-cell but not CFA, promotes the production of high levels of
proliferative responses has been linked to disease severity in infected individuals, IL-10 (REFS 35,36), which, in turn, are important for pro-
which raises the possibility of a role for regulatory T cells in disease modulation. tection against potentially lethal pro-inflammatory
The importance of interleukin-10 in immune-response regulation during mechanisms.
schistosomiasis
is consistent with a role for these cells. What about B cells? The role of B cells in the regulation
• The mechanism that is responsible for naturally acquired immunity. Antibody- of disease is unclear at present (BOX 3). In one report
dependent cellular cytotoxicity (ADCC) that is mediated by immunoglobulin E and (using JH mice), B cells were shown to be essential for
eosinophils is implicated in this process, but definitive proof is lacking. This is a the induction of a TH2 response during infection37.
difficult area, because, for reasons that are outlined in the text, the mouse has been However, in a contrasting study, TH2 responses were
largely discounted as a model for studying naturally acquired immunity. shown to be intact in the absence of B cells (in µMT
• Vaccination. Although some vaccines are in human trials, none promises high levels of mice), but chronic morbidity was markedly enhanced
efficacy. The need exists for the indentification of an effective antigen and/or delivery with grossly enlarged granulomas38; this process was
system for vaccination against schistosomiasis. Fc-receptor-dependent. Animals in which B-cell–T-cell
• The identification of antigens that are involved in the immunopathological response. interactions are compromised by the targeted deletion
of CD80 and CD86 (REF. 39), or CD154 (CD40L)40 also
• Development of T-cell-receptor transgenic mice that are specific for important
schistosome antigens.
fail to develop TH2 responses after infection, although it
is unclear whether B cells are responsible for this effect.
• How schistosomes evade the immune response. This important area of research is in
decline. One of few relevant recent additions to the literature reports that larval
Developmental cues for schistosomes. Given that they
schistosome-derived prostaglandin D2 inhibits the migration of epidermal antigen-
are parasites, it seems logical that schistosomes should
presenting Langerhans cells from the site of infection149, which indicates a way in
be able to use host-derived signals as cues to guide their
which the infection might delay the onset of the immune response. However, the
molecular basis for the prolonged survival of adult schistosomes remains unknown. own development and behaviour41 (BOX 1). Indeed, the
simple observation that, so far, it is impossible to satis-
• Completion of the schistosome genome sequence and life-cycle transcript profiles.
factorily grow schistosomes in tissue culture is evidence
• The development of tools for transgenesis in schistosomes. that these parasites do require very specific signals from
• Schistosome biology as it more broadly relates to the interaction with the host. their hosts42. Attempts to infect mice that have induced
Important issues include understanding the molecular basis of how schistosomes immune-system defects have shown that parasite
interact with immune-system components. Advances in this area will, in part, rely fecundity is markedly reduced in mice with SEVERE
on progress in the preceding two points. COMBINED IMMUNODEFICIENCY (SCID), NUDE mice and T-cell-
depleted mice43,44. These studies indicate that T cells
might be a source of one such host signal. From recent
severe disease, with excessive TH2 responses, and marked studies by Davies and colleagues that examined parasite
mortality during the chronic stages of infection that was development in RAG −/− mice (which lack both T and
associated with increased granuloma size and fibrosis. So, B cells), the reduced parasite fecundity seems to be the
IL-10 might have an important regulatory role in schis- result of a delay in the maturation of parasites in the
tosomiasis, preventing the development of excessive absence of T cells45. Detailed analyses have shown that a
SEVERE COMBINED
IMMUNODEFICIENCY
TH1- and TH2-mediated pathologies. previously unrecognized subset of CD4+ T cells —
(SCID). A condition in which Evidence from studies of S. haematobium-infected which is present in mice that lack both MHC class I
T-cell responses and antibody humans also indicates the importance of IL-10 in regu- and II molecules and is localized primarily within the
production are virtually non- lating morbidity 6,30. The regulation of egg-antigen- liver — is likely to have an important role in promot-
existent, which can be caused by
induced T-cell proliferation in these individuals — for ing schistosome maturation. The exact immunological
several immunological defects.
In mice, this condition is caused whom T-cell proliferation is positively correlated with function of this class of T cells and the mediators that
by the scid mutation. disease severity31 — seems to be under the control of they produce that affect parasite development remain
IL-10 (REF. 32). to be determined. It is possible that the hepatic T cells
NUDE Given the observed severity of disease that is associ- produce, or are dependent on, IL-7, because the pheno-
A mutation in mice that causes
both hairlessness and defective
ated with the excessive development of a TH1 response type that is described for schistosomes in Rag−/− mice is
formation of the thymus, which in Il4−/−Il10 −/− mice, it is notable that mice that were similar to that described for parasites that grow in Il7 −/−
results in a lack of mature T cells. deliberately immunized with schistosome eggs plus mice46. There is ongoing debate about a role for TNF as
IL-12 — to provoke the development of a strong egg- a host factor that can stimulate female schistosomes to
RAG
antigen-specific TH1 response during subsequent produce eggs43,47,48 and, in general, there has been a lack
Recombinase activating genes
(Rag1 and Rag2) are expressed infection — developed less-severe disease than non- of success in defining at a molecular level the interface
in developing lymphocytes. Mice immunized infected mice33, which was characterized by between schistosomes and their host (BOX 3). However,
that are deficient for either of reduced granuloma size and fibrosis. Why do these ani- schistosomes have been found to express a receptor,
these genes fail to produce B or mals not suffer the same fate as infected Il4 −/−Il10 −/− SmRK1, on their surface that can bind the cytokine
T cells owing to a developmental
block in the gene rearrangement
mice? This issue is particularly interesting in view of the transforming growth factor-β (TGF-β), which indi-
that is necessary for receptor recent report that mice that were immunized with egg cates that host cytokines can have effects on these
expression. antigen in complete Freund’s adjuvant (CFA) developed parasites49.

504 | JULY 2002 | VOLUME 2 www.nature.com/reviews/immunol


REVIEWS

Schistosomiasis: effects on concurrent disease Surprisingly, this suppression was found to continue as
Most people who live in areas that are endemic for the TH2 response developed and became dominant.
schistosomiasis are also exposed to many other infec- Overall then, it is difficult to draw clear conclusions
tious diseases. Given the counter-regulatory effects that about alterations in immune responses and associated
are exerted by TH1 and TH2 cells on each other’s devel- changes in disease development in individuals who are
opment, there is growing interest in whether existing infected with HBV or HCV and schistosomiasis.
infection with schistosomes (or any other chronic infec- Moreover, it now seems clear that in Egypt at least, where
tion that is associated with a strongly polarized TH HCV and schistosomiasis are two of the most important
response) influences an individual’s immune response public-health problems and have geographically over-
against, and therefore their susceptibility to, disparate lapping distributions, the coincidence of infection is a
pathogens. In addition, the realization that morbidity result of the unfortunate initial widespread transmission
during schistosomiasis is dependent on the TH2–TH1 of HCV by mass parenteral antischistosomal therapy,
balance of the immune response raises interesting ques- which continued into the 1980s64.
tions about the potential for co-infection to affect the Whether schistosomiasis affects susceptibility to
outcome of pathological changes that are associated HIV-1 or whether these infections interact in any way is
with schistosome infection. an area of much interest at present. In vitro, TH2 cells
IL-4, which is one of the main products of TH2 cells, have, in some cases, been found to support HIV replica-
is important in polarizing TH2 responses50–52. So, in the tion more strongly than TH1 cells65, which led to the
environment that is created by chronic schistosome hypothesis that helminth infections contribute to the
infection, elevated levels of IL-4 might be expected to high prevalence of AIDS and HIV infection in
influence the outcome of immune responses to other Africa66,67. Consistent with the in vitro findings, recent
antigens. Findings that are consistent with this view studies have shown that, compared with T cells in the
have come from the analysis of the outcome of vaccina- peripheral blood of S. mansoni-infected individuals,
tion in schistosome-infected adults and the babies of those in schistosome and HIV co-infected individuals
infected mothers, for which responses to vaccines that responded to egg antigen by making less IL-4 and IL-10,
normally induce TH1 responses (tetanus toxoid and but similar (low) amounts of IFN-γ, which indicates
Mycobacterium bovis bacillus Calmette–Guerin, respec- that there is a swing in the overall balance of the
tively) were found to be significantly impaired53,54. response from TH2 to TH1 (REF. 68). On the basis of the
Similar results have been gained from experimental sys- various roles that have been established for CD4+ T cells
tems using schistosome-infected mice55 and in individ- during schistosomiasis in the mouse model, it would be
uals with other helminth infections56. An important anticipated that individuals with AIDS would have
area of research will be to ascertain whether these types altered patterns of hepatic fibrosis and, perhaps, an
of effect have any impact on individuals that live in increased risk of liver damage owing to the insufficient
schistosome-endemic areas, in terms of vaccine efficacy sequestration of egg hepatotoxins and/or the relatively
or susceptibility to infections that are usually con- increased production of pro-inflammatory cytokines.
trolled by TH1 responses. In experimental settings, In direct contrast to the situations that are discussed
mice that have schistosomiasis are less able to mount above, ongoing TH2 responses in chronic schistosomiasis
specific anti-viral CD8+ and TH1 immune responses might be beneficial during co-infection with other
(and, consequently, are less able to clear virus)57, have pathogens (for example, the intestinal nematode
greater susceptibility to malaria58 and are extremely Trichuris muris 69) against which TH2 responses are host-
susceptible to infection with Toxoplasma gondii59, a protective, and in preventing the onset of TH1-mediated
parasite that induces marked TH1 responses and that is autoimmunity (for example, diabetes mellitus in geneti-
lethal in mice that have defects in IFN-γ production. cally predisposed non-obese diabetic mice70) and miti-
In clinical settings, co-infection of hepatitis B or C gating against allergy. The parallels that exist between the
virus (HBV or HCV) with schistosomes is common immunology of allergy and of helminth infections are
and has been the focus of much attention in the past. obvious, in that both are associated with TH2-dominated
The confluence of these viral and helminth infections immune responses. Paradoxically, allergic disease seems
in the liver, together with the opposite requirements for to be less frequent in developing countries that still have
TH1-like anti-viral immunity and the observed domi- widespread helminth infection71, and evidence is accu-
nant TH2 response during schistosomiasis, offers a pos- mulating that helminth infection might be inversely
sible explanation for the increased occurrence of related to the prevalence of allergy (the ‘hygiene hypoth-
chronic hepatitis-virus infection in schistosomiasis esis’)72,73. Several recent studies in human populations
patients. Indeed, there is evidence that schistosomiasis that are infected with S. mansoni 74 and S. haemato-
prevents the development of TH1 responses to HCV60,61, bium75 have shown a clear inverse relationship between
and some evidence that HBV and HCV infections are a allergen responsiveness and schistosome infection. It
factor in the development of hepatosplenic schistoso- has been proposed that the regulatory mechanisms that
miasis2. However, schistosome infection of HBV- are induced as a component of the immune response to
transgenic mice actually suppressed viral replication in chronic helminth infection, such as the production of
an IFN-γ-dependent manner soon after the onset of IL-10 and, possibly, TGF-β, constitute a pathway by
egg production62 — when IFN-γ and inducible nitric which inflammatory sequelae during allergic responses
oxide synthase (iNOS) are being expressed63 (FIG. 1). might be non-specifically prevented72. Elucidating the

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 5 0 5


REVIEWS

cellular basis of this type of regulatory response will be How do schistosomes induce TH2 responses?
of considerable interest (BOX 3). An inability to make TH2 responses renders mice acutely
sensitive to infection with schistosomes (above) and
Susceptibility factors for infection highly susceptible to intestinal helminth infections86.
In areas where schistosomiasis is endemic, there is an The evolutionary pressure on the immune system to
obvious pattern of age-dependent intensity of infec- recognize helminths as pathogens against which TH2
tion; individuals who are below the age of puberty responses should be made is, therefore, apparent.
carry the most parasites, and those in older age brack- However, the mechanisms by which the immune system
ets are generally less heavily infected76. Drug treatment accomplishes this are unclear, and they are a subject of
of affected populations followed by careful assessment intense interest (FIG. 3).
of reinfection status has shown that children usually It has been recognized for some time that it is the egg
become heavily reinfected, whereas older individuals stage of the schistosome that is responsible for inducing
might become reinfected, but remain less heavily the TH2 response during infection87,88. By contrast, the
infected than they were before treatment. So, in worms themselves seem to be poor inducers of a TH2
endemic areas, older individuals are resistant to re- response. As for certain other helminth products89, schis-
infection. A comparison of immune responses tosome eggs or soluble antigens that are derived from the
between those individuals who are susceptible and eggs induce an intense TH2 response without the need
those who are resistant to reinfection has shown that for additional adjuvant87,90. Recent work has shown that
there is a correlation between immunoglobulin-E carbohydrates on egg antigens are integral to this
responses to worm (not egg) antigens and immunity, process91,92 and, specifically, that a polylactosamine sugar
which implicates IgE in the protective effector mecha- (lacto-N-fucopentaose III) acts as a TH2 adjuvant93. The
nism77–80. The slow development of appropriate emerging role of carbohydrates as factors that are impor-
immune responses to worm antigens might be linked tant for the induction of the immune response during
to the fact that schistosomes are very long-lived para- schistosomiasis opens up the possibility that innate
sites and the host becomes exposed to these antigens pattern-recognition receptors that identify carbohy-
only after parasites die81, either as a result of ageing or drates might have a crucial role in the induction of a
drug intervention. Consistent with the immuno- TH2 response. The recent identification of a wide range
epidemiological data, results from studies in Brazil of C-type lectin receptors that are expressed on the sur-
have shown that the intensity of infection is influenced face of dendritic cells (DCs)94 indicates various candi-
by a major gene (SM1) that maps to a region of chro- dates that could be involved in the innate recognition of
mosome 5 (5q31–q33) that encodes the TH2 antigens from pathogens that initiate a TH2 response.
cytokines82. How IgE functions in a protective capacity It remains to be seen whether Toll-like receptors
in people is unclear, but interaction with eosinophils in (TLRs) — which have crucial roles in the recognition of
an antibody-dependent cellular cytotoxicity (ADCC) viral, bacterial, fungal and protozoal organisms, and in
reaction that is targeted at schistosomula is a favoured the development of TH1 immune responses95 — exist
model83,84 (BOX 3). for the recognition of helminth antigens and/or have
Mice that are infected with S. mansoni are unable any role in the induction of TH2 responses. This would
to clear the primary infection, but nevertheless are provide an attractive innate mechanism to allow appro-
partially resistant to superinfection. However, the use priate immune-response development to the wide
of mice for studies of resistance to reinfection has diversity of helminth parasites. However, no specific
been questioned on two points. First, resistance in pattern-recognition receptors have been identified for
mice might, in large part, be due to the development this role so far. Indeed, current data indicate that sig-
of portosystemic vascular shunts85 (BOX 2). In these nalling through MYD88, which is the main adaptor
animals, immature parasites of a secondary infection protein for the known TLRs, is not necessary for the
might find it difficult to localize to the portal vascula- development of TH2 responses96.
ture and, instead, will be carried by the blood flow, Directly related to these areas, recent data indicate
through varices, to non-permissive areas of the vascu- that schistosomes and other helminths might affect
lature. This resistance is, therefore, more anatomical TH2-response development by influencing the way in
than immunological, and it is related to pathological which DCs become activated97–99. Helminth antigens, in
changes that are more prevalent in infected mice than contrast to most microbial pathogens, seem not to ‘clas-
in infected humans2. Second, the cellular distribution sically’ activate DCs; in particular, they fail to induce the
of the high-affinity receptor for IgE (FcεR1) on mouse production of IL-12 by DCs. Furthermore, mouse DCs
cells differs from that on human cells 84. As IgE- that are exposed to schistosome egg antigens also fail to
dependent eosinophil-mediated ADCC is a possible upregulate their expression of surface markers that are
effector mechanism of protective immunity in normally associated with the activation or maturation
humans, the lack of FcεR1 on mouse eosinophils is of of DCs98. However, DCs that are exposed to egg anti-
particular concern when attempting to model human gens are strong inducers of TH2 cells both in vitro and
immunity using the mouse 84. One consequence of in vivo98. The mechanism by which DCs induce TH2
this is the current lack of definitive data to indicate an responses has been a somewhat neglected area of study,
in vivo role for eosinophils in any immunological and, so far, it remains poorly defined100. An attractive
process during schistosomiasis (BOX 3). possibility, for which some supporting data exist101, is

506 | JULY 2002 | VOLUME 2 www.nature.com/reviews/immunol


REVIEWS

Periphery Lymphoid
tissue
Glycoproteins

?
Immature IL-12
dendritic cell
No
No n-clas
IL-1 sica
Eggs 2 p l ac IL-10
rod tiva TH 1
uct tion IFN-γ
Lectin ? ion
TLR ? Mig
rati ?
on
Secreted
antigens Additonal
signals ?

Dendritic
cell T cell
IL-4 ?

TH 2
TH 2
MHC OX40L CD40
class II IL-4
TH 2
IL-5
IL-13

TCR OX40 CD154

Figure 3 | TH2-response induction by schistosome egg antigens. Immature dendritic cells (DCs) can acquire schistosome egg
antigens and induce T helper 2 (TH2) responses, but the process by which this occurs is unclear. This figure shows one possible
model. Many egg proteins are glycosylated, and carbohydrates are implicated in the induction of TH2 responses by these antigens;
possibly, DCs acquire egg antigens through lectin-like receptors. The exposure of DCs to egg antigens does not result in the
classical activation changes that are described for DCs that are exposed to lipopolysaccharide or Gram-positive bacteria; they do
not make interleukin-12 (IL-12), and they do not upregulate their expression of the co-stimulatory molecules CD40, CD80 or CD86.
Nevertheless, such DCs are able to initiate immune responses, presumably by migrating to lymphoid organs, where they encounter
and activate naive CD4+ T cells. Development of the TH2 response is dependent on IL-4 from a source other than the DC that is
inducing the response. It is possible that IL-10 that is produced by the DC has a role in suppressing IL-12 production and minimizing
the progression of the TH1 response. IL-4 would also be expected to limit TH1-response development and to act as a growth factor
to expand the TH2 response. There is a clear role for CD40–CD154 interactions in TH2-response development during
schistosomiasis; CD80 and CD86 also seem to be important (not shown) and OX40L–OX40 interactions have been implicated in
the development of TH2 responses to schistosome antigens99. IFN-γ, interferon-γ; TCR, T-cell receptor; TLR, Toll-like receptor.

that TH2-response-inducing pathogens stimulate DCs Several cytokines other than IL-4 have been impli-
to produce IL-4, which then promotes TH2-response cated in TH2 development, but, on closer examination,
development (FIG. 3). However, it is now clear that DCs have been found to be of minimal importance for the
do not need to produce IL-4 to direct TH2 develop- expression of this type of immune response during
ment102, because egg-antigen-pulsed Il4 −/− DCs induce schistosomiasis. IL-13, which is closely related to IL-4,
excellent TH2 responses when injected into naive mice, seems to be crucial for granuloma formation and fibro-
so long as the recipient animal is able to make IL-4. sis (see above), but not to be necessary for TH2 develop-
Although IL-4 has an important role in this model of ment per se 10,14. IL-6 can direct the development of
egg-antigen-induced TH2 responses, and is important IL-4-producing T cells104. However, IL-6 does not have a
for the normal development of TH2 responses during main role during the development of TH2 responses to
schistosomiasis (see above), recent evidence indicates schistosome eggs in vivo105, although it might be
that neither IL-4R nor the downstream signal trans- involved at some level in the regulation of IFN-γ and
ducer and activator of transcription 6 (STAT6) are IL-12 production106. The role of IL-10 in TH2-response
absolutely required103, because infection of mice that consolidation has been discussed extensively above.
are deficient in these molecules results in small, but The recent description of effector B cells that can
measurable, TH2 responses. induce TH2 responses through the production of

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 5 0 7


REVIEWS

polarizing cytokines107 raises the possibility, supported by protective immunity to schistosomes is the radiation-
one study37, that B cells might have a role in the establish- attenuated cercarial vaccine, which is able to induce
ment or maintenance of TH2 responses in schistosome- consistently high, although not sterilizing, immunity
infected mice. The finding that CD40–CD154 interac- against challenge infection in mice121,122. A single expo-
tions are important for TH2-response development sure to irradiated cercariae induces a TH1 response,
during schistosomiasis40,108 implicates B cells in this whereas additional boosting leads to a mixed TH1/TH2
process, because B-cell responses are markedly impaired response123. In mice that are immunized a single time,
in the absence of CD40 signalling109. In addition, it has the protective effector mechanism seems to be
become apparent recently that CD40 −/− DCs are inca- cell mediated, and IFN-γ- and/or TNF-activated
pable of inducing egg-antigen-specific TH2 responses110. macrophages and NO are implicated124–127; consistent
Together, these data indicate that the CD40–CD154 with this, IL-12 and bacterial CpG motifs (which
interaction is required for egg-antigen-induced TH2 induce the production of IL-12 by DCs and
responses, but they leave open the question of the role of macrophages that express the appropriate TLR95) can
B cells in this process. be used as adjuvants to boost immunity128,129. However,
In addition to CD40–CD154, several members of the an underlying protective B-cell antibody-dependent
B7 superfamily111 have been investigated in terms of mechanism also exists in animals that are vaccinated in
their influence on TH2-response induction by schisto- this way127,130. In mice that have been vaccinated many
somes. Mice that are doubly deficient for both CD80 times, antibodies begin to have a more important pro-
and CD86 fail to mount a TH2 response to schistosome tective role131. Optimal protection in all cases is proba-
infection and have an impaired ability to form granulo- bly linked to the induction of cell-mediated and
mas39. CD86 is probably more important than CD80 in humoral responses122,127. Consistent with the develop-
this regard, because the infection of mice that are singly ment of exaggerated TH1 and TH2 responses in infected
deficient for CD86, but not for CD80, results in reduced Il10 −/− mice, vaccinated Il10 −/− mice also develop
egg-antigen-specific proliferation and TH2 cytokine pro- enhanced immune responses with integral TH1 and TH2
duction by cultured T cells from infected mice39. components, and are almost entirely resistant to infec-
Furthermore, although anti-CD80 antibodies have no tion132, which indicates that a high-magnitude multi-
effect on transcript levels for IL-4 or IL-5 in a pulmonary faceted immune response might be the best option for
model of schistosome egg-induced granuloma forma- induced resistance to schistosome infection. However,
tion, treatment with anti-CD86 antibody inhibits the the fact remains that after many years of trying, a ratio-
expression of IL-4, IL-5 and IL-13 (REF. 112). Two other nally designed effective anti-schistosome subunit vac-
molecules that have been implicated recently in the cine has yet to be developed. To obtain an effective
induction of a TH2 response are inducible co-stimulator vaccine against such a complex and highly successful
(ICOS)113 and the IL-1R-related molecule T1/ST2 metazoan pathogen is a daunting challenge.
(REF. 114). However, blockade of the interaction of ICOS Nevertheless, the dangers that are presented by the pos-
with its ligand, B7-related protein 1 (B7RP1), using a sible emergence of drug-resistant schistosomes133, cou-
soluble ICOS fusion protein, did not affect TH2-cell dif- pled with the fact that the prevalence of schistosomiasis
ferentiation in a model of allergic airway inflammation has remained the same since effective orally adminis-
that used S. mansoni eggs as the antigenic stimulus115. tered chemotherapy became widely available, demand
The expression of T1/ST2 seems to be enhanced on continued and active research in this area (BOX 3).
CD4+ T cells that are isolated from schistosome egg- An alternative approach to vaccination, as discussed
induced lung granulomas or from the livers of infected above, is to induce a TH1-like immune response that
mice116. Furthermore, the expression of T1/ST2 has been can prevent the normal TH1 to TH2 transition that
implicated in TH2 cytokine production ex vivo after the occurs in infected hosts after the onset of egg produc-
intravenous injection of S. mansoni eggs117. It remains to tion by the parasites, with the aim of preventing
be determined how important either T1/ST2 or ICOS the development of severe chronic morbidity134.
might be for TH2-response development during actual Experimentally, this approach has been promising.
schistosome infection. However, there are legitimate concerns about the
So, a wide range of both parasite- and host-derived potential for exaggerated early-onset liver disease that
candidates have been assessed for their role in the induc- is associated with this type of induced immune devia-
tion of a TH2 response by schistosomes. Determining tion34,135. Moreover, it is not clear what effect predispos-
exactly how these many components might fit together ing individuals to make TH1 responses would have on
is now crucial. A pressing issue is whether TH2-response their ability to subsequently develop resistance to infec-
development occurs simply as a default pathway when tion, given that the latter seems to be TH2-response-
IL-12 and related mediators are not induced. mediated in the endemic setting. Nevertheless, the
philosophy of vaccinating to prevent disease, rather
Vaccine-induced immunity than infection, remains enticing, and the approach
Several detailed reviews of the primary candidate vaccines becomes more plausible as its immunological basis is
for schistosomiasis have been published recently118–120. increasingly well understood16. It remains to be seen
Consequently, in this review, we focus on the immuno- whether this type of vaccination can be reproduced
logical mechanisms that underlie vaccine-induced using defined antigens that are amenable to large-scale
immunity. The basis for many of the developments in manufacture.

508 | JULY 2002 | VOLUME 2 www.nature.com/reviews/immunol


REVIEWS

Conclusions and future directions this balance is achieved. This information, in turn,
Schistosomes are remarkable metazoan parasites that might help in the design of rational therapeutics for
have co-evolved in concert with their mammalian schistosomiasis and related diseases. Understanding
hosts such that they are dependent on certain immune- how the host makes the decision to mount TH2
system components for their own biology. The responses during schistosomiasis remains an area of
immune system is largely incapable of resisting primary high priority, as does understanding the impact of the
infection, and resistance to superinfection takes years to immune response that is induced by schistosome infec-
develop. So, the survival of the host seems to depend on tion on the outcome of immunity to other pathogens
the ability to make an appropriately balanced TH (and vice versa). Renewed effort to understand the
response that is able to orchestrate granuloma develop- basic biology of schistosomes, including their immune-
ment, prevent debilitating acute disease, and minimize evasion mechanisms136, is clearly necessary, as this
fibrosis and severe morbidity during chronic infection. remains poorly understood137. More detailed informa-
Amazingly, most (>90%) infected individuals in tion about these areas, coupled with a high-resolution
endemic areas seem to successfully accomplish this. view of the type of immune response that is necessary
Future work using DNA microarray analyses and for resistance to infection with schistosomes, will
refined genetics promises to reveal much about how facilitate vaccine design.

1. van der Werf, M. J. et al. Quantification of clinical morbidity balance in mouse bone-marrow-derived macrophages by immunopathology in murine schistosomiasis. J. Immunol.
associated with schistosome infection in sub-Saharan TH1 and TH2 cytokines. Eur. J. Immunol. 25, 1101–1104 164, 6406–6416 (2000).
Africa. Acta Tropica (in the press). (1995). This study establishes the immunological
A comprehensive assessment of the true impact of 16. Hesse, M., Cheever, A. W., Jankovic, D. & Wynn, T. A. NOS-2 requirements for minimizing disease during the acute
schistosomiasis on human health. mediates the protective anti-inflammatory and antifibrotic and chronic phases of schistosomiasis.
2. Dunne, D. W. & Pearce, E. J. Immunology of hepatosplenic effects of the TH1-inducing adjuvant, IL-12, in a TH2 model of 29. Vaillant, B., Chiaramonte, M. G., Cheever, A. W., Soloway, P. D.
schistosomiasis mansoni: a human perspective. Microbes granulomatous disease. Am. J. Pathol. 157, 945–955 & Wynn, T. A. Regulation of hepatic fibrosis and extracellular
Infect. 1, 553–560 (1999). (2000). matrix genes by the TH response: new insight into the role of
3. Cheever, A. W., Hoffmann, K. F. & Wynn, T. A. 17. Lee, C. G. et al. Interleukin-13 induces tissue fibrosis by tissue inhibitors of matrix metalloproteinases. J. Immunol.
Immunopathology of schistosomiasis mansoni in mice and selectively stimulating and activating transforming growth 167, 7017–7026 (2001).
men. Immunol. Today 21, 465–466 (2000). factor-β1. J. Exp. Med. 194, 809–821 (2001). 30. King, C. L. et al. Schistosoma haematobium-induced
4. Rabello, A. Acute human schistosomiasis mansoni. Mem. 18. Hesse, M. et al. Differential regulation of nitric oxide urinary-tract morbidity correlates with increased tumor-
Inst. Oswaldo Cruz 90, 277–280 (1995). synthase-2 and arginase-1 by type 1/type 2 cytokines necrosis factor-α and diminished interleukin-10 production.
5. de Jesus, A. R. et al. Clinical and immunologic evaluation of in vivo: granulomatous pathology is shaped by the pattern of J. Infect. Dis. 184, 1176–1182 (2001).
31 patients with acute schistosomiasis mansoni. J. Infect. L-arginine metabolism. J. Immunol. 167, 6533–6544 (2001). 31. Colley, D. G. In Idiotypic Network and Diseases (eds Cerney, J.
Dis. 185, 98–105 (2002). 19. Mohamed-Ali, Q. et al. Susceptibility to periportal (Symmers) & Hiernauz, J.) 71–105 (American Society for Microbiology,
6. Montenegro, S. M. et al. Cytokine production in acute fibrosis in human Schistosoma mansoni infections: evidence Washington DC, 1990).
versus chronic human schistosomiasis mansoni: the cross- that intensity and duration of infection, gender and inherited 32. King, C. L. et al. Cytokine control of parasite-specific anergy
regulatory role of interferon-γ and interleukin-10 in the factors are critical in disease progression. J. Infect. Dis. 180, in human urinary schistosomiasis. IL-10 modulates
responses of peripheral blood mononuclear cells and 1298–1306 (1999). lymphocyte reactivity. J. Immunol. 156, 4715–4721 (1996).
splenocytes to parasite antigens. J. Infect. Dis. 179, 20. Dessein, A. J. et al. Severe hepatic fibrosis in Schistosoma 33. Wynn, T. A. et al. An IL-12-based vaccination method for
1502–1514 (1999). mansoni infection is controlled by a major locus that is preventing fibrosis induced by schistosome infection. Nature
7. King, C. L. et al. B-cell sensitization to helminthic infection closely linked to the interferon-γ receptor gene. Am. J. Hum. 376, 594–596 (1995).
develops in utero in humans. J. Immunol. 160, 3578–3584 Genet. 65, 709–721 (1999). 34. Rutitzky, L. I., Hernandez, H. J. & Stadecker, M. J. TH1-
(1998). Severe schistosomiasis occurs in less than 10% of polarizing immunization with egg antigens correlates with
8. Malhotra, I. et al. In utero exposure to helminth and infected individuals. This report is an important step
severe exacerbation of immunopathology and death in
mycobacterial antigens generates cytokine responses towards understanding the genetic predispostion to
schistosome infection. Proc. Natl Acad. Sci. USA 98,
similar to that observed in adults. J. Clin. Invest. 99, severe disease.
13243–13248 (2001).
1759–1766 (1997). 21. Araujo, M. I. et al. Evidence of a T helper type-2 activation in
References 33 and 34 show that induced TH1
9. Brunet, L. R., Finkelman, F. D., Cheever, A. W., Kopf, M. A. & human schistosomiasis. Eur. J. Immunol. 26, 1399–1403
responses against egg antigens can lead to reduced
Pearce, E. J. IL-4 protects against TNF-α-mediated (1996).
hepatic fibrosis, but that there is a risk of severe
cachexia and death during acute schistosomiasis. 22. Williams, M. E. et al. Leukocytes of patients with
disease in mice that are immunologically polarized in
J. Immunol. 159, 777–785 (1997). Schistosoma mansoni respond with a TH2 pattern of
this way. These papers emphasize the importance of
The first paper to show that the TH2 response that is cytokine production to mitogen or egg antigens, but with a
the appropriate immunological balance for optimal
induced during schistosomiasis is essential for host TH0 pattern to worm antigens. J. Infect. Dis. 170, 946–954
outcome during infection.
survival. (1994).
35. Wynn, T. A., Eltoum, I., Oswald, I. P., Cheever, A. W. & Sher, A.
10. Fallon, P. G., Richardson, E. J., McKenzie, G. J. & 23. Mwatha, J. K. et al. High levels of TNF, soluble TNF
Endogenous interleukin-12 (IL-12) regulates granuloma
McKenzie, A. N. Schistosome infection of transgenic mice receptors, soluble ICAM-1, and IFN-γ, but low levels of IL-5,
defines distinct and contrasting pathogenic roles for IL-4 are associated with hepatosplenic disease in human formation induced by eggs of Schistosoma mansoni, and
and IL-13: IL-13 is a profibrotic agent. J. Immunol. 164, schistosomiasis mansoni. J. Immunol. 160, 1992–1999 exogenous IL-12 both inhibits and prophylactically
2585–2591 (2000). (1998). immunizes against egg pathology. J. Exp. Med. 179,
11. La Flamme, A. C., Patton, E. A., Bauman, B. & Pearce, E. J. 24. Dessein, A. J. et al. Infection and disease in human 1551–1561 (1994).
IL-4 plays a crucial role in regulating oxidative damage in the schistosomiasis mansoni are under distinct major gene 36. Morris, S. C. et al. Effects of IL-12 on in vivo cytokine gene
liver during schistosomiasis. J. Immunol. 166, 1903–1911 control. Microbes Infect. 1, 561–567 (1999). expression and Ig-isotype selection. J. Immunol. 152,
(2001). 25. Montesano, M. A., Colley, D. G., Willard, M. T., Freeman, 1047–1056 (1994).
12. Hatz, C. F. The use of ultrasound in schistosomiasis. Adv. G. L. Jr & Secor, W. E. Idiotypes expressed early in 37. Hernandez, H. J., Wang, Y. & Stadecker, M. J. In infection
Parasitol. 48, 225–284 (2001). experimental Schistosoma mansoni infections predict with Schistosoma mansoni, B cells are required for T helper
13. Jankovic, D. et al. Schistosome-infected IL-4 receptor clinical outcomes of chronic disease. J. Exp. Med. 195, type-2 cell responses but not for granuloma formation.
knockout (KO) mice, in contrast to IL-4 KO mice, fail to 1223–1228 (2002). J. Immunol. 158, 4832–4837 (1997).
develop granulomatous pathology while maintaining the 26. Bosshardt, S. C., Freeman, G. L. Jr, Secor, W. E. & 38. Jankovic, D. et al. CD4+ T-cell-mediated granulomatous
same lymphokine expression profile. J. Immunol. 163, Colley, D. G. IL-10 deficit correlates with chronic, pathology in schistosomiasis is downregulated by a B-cell-
337–342 (1999). hypersplenomegaly syndrome in male CBA/J mice infected dependent mechanism requiring Fc receptor signaling.
14. Chiaramonte, M. G., Donaldson, D. D., Cheever, A. W. & with Schistosoma mansoni. Parasite Immunol. 19, 347–353 J. Exp. Med. 187, 619–629 (1998).
Wynn, T. A. An IL-13 inhibitor blocks the development of (1997). 39. Hernandez, H. J., Sharpe, A. H. & Stadecker, M. J.
hepatic fibrosis during a T-helper type-2-dominated 27. Montesano, M. A., Colley, D. G., Eloi-Santos, S., Freeman, Experimental murine schistosomiasis in the absence of B7
inflammatory response. J. Clin. Invest. 104, 777–785 (1999). G. L. Jr & Secor, W. E. Neonatal idiotypic exposure alters costimulatory molecules: reversal of elicited T-cell cytokine
This report established IL-13 as a profibrogenic subsequent cytokine, pathology and survival patterns in profile and partial inhibition of egg granuloma formation.
mediator in schistosomiasis, and it describes a experimental Schistosoma mansoni infections. J. Exp. Med. J. Immunol. 162, 2884–2889 (1999).
rationally designed experimental immunotherapy that 189, 637–645 (1999). 40. MacDonald, A. S. et al. Impaired TH2 development and
blocks fibrosis. 28. Hoffmann, K. F., Cheever, A. W. & Wynn, T. A. IL-10 and increased mortality during Schistosoma mansoni infection
15. Modolell, M., Corraliza, I. M., Link, F., Soler, G. & Eichmann, K. the dangers of immune polarization: excessive type 1 and in the absence of CD40/CD154 interaction. J. Immunol.
Reciprocal regulation of the nitric oxide synthase/arginase type 2 cytokine responses induce distinct forms of lethal 168, 4643–4649 (2002).

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 5 0 9


REVIEWS

41. Salzet, M., Capron, A. & Stefano, G. B. Molecular crosstalk 64. Frank, C. et al. The role of parenteral antischistosomal schistosomiasis leads to the development of a strong
in host–parasite relationships: schistosome– and leech–host therapy in the spread of hepatitis C virus in Egypt. Lancet TH2 response.
interactions. Parasitol. Today 16, 536–540 (2000). 355, 887–891 (2000). 89. Holland, M. J., Harcus, Y. M., Riches, P. L. & Maizels, R. M.
42. Basch, P. F. & Rhine, W. D. Schistosoma mansoni: 65. Maggi, E. et al. Ability of HIV to promote a TH1 to TH0 shift Proteins secreted by the parasitic nematode
reproductive potential of male and female worms cultured and to replicate preferentially in TH2 and TH0 cells. Science Nippostrongylus brasiliensis act as adjuvants for TH2
in vitro. J. Parasitol. 69, 567–569 (1983). 265, 244–248 (1994). responses. Eur. J. Immunol. 30, 1977–1987 (2000).
43. Amiri, P. et al. Tumour-necrosis factor-α restores granulomas 66. Bentwich, Z., Kalinkovich, A. & Weisman, Z. Immune 90. Vella, A. T. & Pearce, E. J. CD4+ TH2 response induced by
and induces parasite egg-laying in schistosome-infected activation is a dominant factor in the pathogenesis of African Schistosoma mansoni eggs develops rapidly, through an
SCID mice. Nature 356, 604–607 (1992). AIDS. Immunol. Today 16, 187–191 (1995). early, transient, TH0-like stage. J. Immunol. 148, 2283–2290
44. Harrison, R. A. & Doenhoff, M. J. Retarded development of 67. Bentwich, Z. et al. Can eradication of helminthic infections (1992).
Schistosoma mansoni in immunosuppressed mice. change the face of AIDS and tuberculosis? Immunol. Today 91. Okano, M., Satoskar, A. R., Nishizaki, K., Abe, M. & Harn,
Parasitology 86, 429–438 (1983). 20, 485–487 (1999). D. A. Jr. Induction of TH2 responses and IgE is largely due to
45. Davies, S. J. et al. Modulation of blood-fluke development in 68. Mwinzi, P. N., Karanja, D. M., Colley, D. G., Orago, A. S. & carbohydrates functioning as adjuvants on Schistosoma
the liver by hepatic CD4+ lymphocytes. Science 294, Secor, W. E. Cellular immune responses of schistosomiasis mansoni egg antigens. J. Immunol. 163, 6712–6717 (1999).
1358–1361 (2001). patients are altered by human immunodeficiency virus type 1 The first report that carbohydrates on egg antigens
This study expands on previous reports that coinfection. J. Infect. Dis. 184, 488–496 (2001). are important for the induction of TH2 responses. See
schistosomes fail to develop properly in hosts that 69. Curry, A. J. et al. Evidence that cytokine-mediated immune also reference 93.
lack T cells. It raises many unanswered questions interactions induced by Schistosoma mansoni alter disease 92. Williams, D. L., Asahi, H., Botkin, D. J. & Stadecker, M. J.
about the role of the previously unidentified CD4+ outcome in mice concurrently infected with Trichuris muris. Schistosome infection stimulates host CD4+ T helper cell and
subset of hepatic lymphocytes that seems to have an J. Exp. Med. 181, 769–774 (1995). B-cell responses against a novel egg antigen, thioredoxin
important role in this process, and the identity of the 70. Cooke, A. et al. Infection with Schistosoma mansoni peroxidase. Infect. Immun. 69, 1134–1141 (2001).
mediator they produce that is used by the parasites. prevents insulin-dependent diabetes mellitus in non-obese 93. Okano, M., Satoskar, A. R., Nishizaki, K. & Harn, D. A. Jr.
46. Wolowczuk, I. et al. Infection of mice lacking interleukin-7 diabetic mice. Parasite Immunol. 21, 169–176 (1999). Lacto-N-fucopentaose III found on Schistosoma mansoni
(IL-7) reveals an unexpected role for IL-7 in the development 71. Worldwide variation in prevalence of symptoms of asthma, egg antigens functions as adjuvant for proteins by inducing
of the parasite Schistosoma mansoni. Infect. Immun. 67, allergic rhinoconjunctivitis and atopic eczema: ISAAC. The TH2-type response. J. Immunol. 167, 442–450 (2001).
4183–4190 (1999). International Study of Asthma and Allergies in Childhood 94. Figdor, C. G., van Kooyk, Y. & Adema, G. J. C-type lectin
47. Cheever, A. W., Poindexter, R. W. & Wynn, T. A. Egg laying is (ISAAC) Steering Committee. Lancet 351, 1225–1232 receptors on dendritic cells and langerhans cells. Nature
delayed but worm fecundity is normal in SCID mice infected (1998). Rev. Immunol. 2, 77–84 (2002).
with Schistosoma japonicum and S. mansoni with or 72. Yazdanbakhsh, M., Kremsner, P. G. & van Ree, R. Allergy, 95. Akira, S., Takeda, K. & Kaisho, T. Toll-like receptors: critical
without recombinant tumor-necrosis factor-α treatment. parasites and the hygiene hypothesis. Science 296, proteins linking innate and acquired immunity. Nature
Infect. Immun. 67, 2201–2208 (1999). 490–494 (2002). Immunol. 2, 675–680 (2001).
48. Davies, S. J. & McKerrow, J. H. In Biology of Parasitism (eds 73. Wills-Karp, M., Santeli, J. & Karp, C. L. The germless theory 96. Schnare, M. et al. Toll-like receptors control activation of
Tschudi, C. & Pearce, E. J.) 273–290 (Kluwer, Boston, of allergic disease: revisiting the hygiene hypothesis. Nature adaptive immune responses. Nature Immunol. 2, 947–950
2001). Rev. Immunol. 1, 69–74 (2001). (2001).
49. Beall, M. J. & Pearce, E. J. Human transforming growth 74. Araujo, M. I. et al. Inverse association between skin 97. Whelan, M. et al. A filarial nematode-secreted product
factor-β activates a receptor serine/threonine kinase from response to aeroallergens and Schistosoma mansoni signals dendritic cells to acquire a phenotype that drives
the intravascular parasite Schistosoma mansoni. J. Biol. infection. Int. Arch. Allergy Immunol. 123, 145–148 (2000). development of TH2 cells. J. Immunol. 164, 6453–6460
Chem. 276, 31613–31619 (2001). 75. van den Biggelaar, A. H. et al. Decreased atopy in children (2000).
50. Murphy, K. M. T-lymphocyte differentiation in the periphery. infected with Schistosoma haematobium: a role for parasite- 98. MacDonald, A. S., Straw, A. D., Bauman, B. & Pearce, E. J.
Curr. Opin. Immunol. 10, 226–232 (1998). induced interleukin-10. Lancet 356, 1723–1727 (2000). CD8− dendritic-cell activation status plays an integral role in
51. Ouyang, W. et al. Stat6-independent GATA-3 autoactivation The identification of IL-10 as an important regulator of influencing TH2 response development. J. Immunol. 167,
directs IL-4-independent TH2 development and allergic manifestations in schistosomiasis. This area 1982–1988 (2001).
commitment. Immunity 12, 27–37 (2000). is reviewed in detail in reference 72. 99. de Jong, E. C. et al. Microbial compounds selectively induce
52. Ouyang, W. et al. Inhibition of TH1 development mediated by 76. Butterworth, A. E. et al. Immunity and morbidity in human TH1-cell-promoting or TH2-cell-promoting dendritic cells
GATA-3 through an IL-4-independent mechanism. Immunity schistosomiasis mansoni. Trop. Geogr. Med. 46, 197–208 in vitro with diverse TH-cell-polarizing signals. J. Immunol.
9, 745–755 (1998). (1994). 168, 1704–1709 (2002).
53. Sabin, E. A., Araujo, M. I., Carvalho, E. M. & Pearce, E. J. 77. Dunne, D. W. et al. Immunity after treatment of human 100. Kalinski, P., Hilkens, C. M., Wierenga, E. A. & Kapsenberg,
Impairment of tetanus toxoid-specific TH1-like immune schistosomiasis: association between IgE antibodies to M. L. T-cell priming by type-1 and type-2 polarized dendritic
responses in humans infected with Schistosoma mansoni. adult worm antigens and resistance to reinfection. Eur. J. cells: the concept of a third signal. Immunol. Today 20,
J. Infect. Dis. 173, 269–272 (1996). Immunol. 22, 1483–1494 (1992). 561–567 (1999).
54. Malhotra, I. et al. Helminth- and Bacillus Calmette-Guerin- 78. Demeure, C. E. et al. Resistance to Schistosoma mansoni in 101. d’Ostiani, C. F. et al. Dendritic cells discriminate between
induced immunity in children sensitized in utero to filariasis humans: influence of the IgE/IgG4 balance and IgG2 in yeasts and hyphae of the fungus Candida albicans.
and schistosomiasis. J. Immunol. 162, 6843–6848 (1999). immunity to reinfection after chemotherapy. J. Infect. Dis. Implications for initiation of T helper cell immunity in vitro and
55. Kullberg, M. C., Pearce, E. J., Hieny, S. E., Sher, A. & 168, 1000–1008 (1993). in vivo. J. Exp. Med. 191, 1661–1674 (2000).
Berzofsky, J. A. Infection with Schistosoma mansoni alters 79. Rihet, P., Demeure, C. E., Bourgois, A., Prata, A. & Dessein, 102. MacDonald, A. S. & Pearce, E. J. Cutting edge: polarized
TH1/TH2 cytokine responses to a non-parasite antigen. A. J. Evidence for an association between human resistance TH-cell response induction by transferred antigen-pulsed
J. Immunol. 148, 3264–3270 (1992). to Schistosoma mansoni and high anti-larval IgE levels. Eur. dendritic cells is dependent on IL-4 or IL-12 production by
56. Cooper, P. J., Espinel, I., Paredes, W., Guderian, R. H. & J. Immunol. 21, 2679–2686 (1991). recipient cells. J. Immunol. 168, 3127–3130 (2002).
Nutman, T. B. Impaired tetanus-specific cellular and humoral 80. Hagan, P., Blumenthal, U. J., Dunn, D., Simpson, A. J. & A clear demonstration that dendritic cells can interpret
responses following tetanus vaccination in human Wilkins, H. A. Human IgE, IgG4 and resistance to reinfection pathogen-inherent signals and drive egg-antigen-
onchocerciasis: a possible role for interleukin-10. J. Infect. with Schistosoma haematobium. Nature 349, 243–245 specific TH2 responses independent of any requirement
Dis. 178, 1133–1138 (1998). (1991). to make IL-4. See also references 97 and 98.
57. Actor, J. K. et al. Helminth infection results in decreased 81. Woolhouse, M. E. & Hagan, P. Seeking the ghost of worms 103. Jankovic, D. et al. Single-cell analysis reveals that IL-4
virus-specific CD8+ cytotoxic T-cell and TH1 cytokine past. Nature Med. 5, 1225–1227 (1999). receptor/Stat6 signaling is not required for the in vivo or in
responses, as well as delayed virus clearance. Proc. Natl 82. Marquet, S. et al. Genetic localization of a locus controlling vitro development of CD4+ lymphocytes with a TH2 cytokine
Acad. Sci. USA 90, 948–952 (1993). the intensity of infection by Schistosoma mansoni on profile. J. Immunol. 164, 3047–3055 (2000).
58. Helmby, H., Kullberg, M. & Troye-Blomberg, M. Altered chromosome 5q31–q33. Nature Genet. 14, 181–184 104. Rincon, M., Anguita, J., Nakamura, T., Fikrig, E. & Flavell, R. A.
immune responses in mice with concomitant Schistosoma (1996). Interleukin (IL)-6 directs the differentiation of IL-4-producing
mansoni and Plasmodium chabaudi infections. Infect. 83. Nutten, S. et al. From allergy to schistosomes: role of Fc CD4+ T cells. J. Exp. Med. 185, 461–469 (1997).
Immun. 66, 5167–5174 (1998). receptors and adhesion molecules in eosinophil effector 105. La Flamme, A. C. & Pearce, E. J. The absence of IL-6 does
59. Marshall, A. J. et al. Toxoplasma gondii and Schistosoma function. Mem. Inst. Oswaldo Cruz 92 (Suppl. 2), 9–14 not affect TH2-cell development in vivo, but does lead to
mansoni synergize to promote hepatocyte dysfunction (1997). impaired proliferation, IL-2 receptor expression and B-cell
associated with high levels of plasma TNF-α and early death 84. Dombrowicz, D. & Capron, M. Eosinophils, allergy and responses. J. Immunol. 162, 5829–5837 (1999).
in C57BL/6 mice. J. Immunol. 163, 2089–2097 (1999). parasites. Curr. Opin. Immunol. 13, 716–720 (2001). 106. La Flamme, A. C., MacDonald, A. S. & Pearce, E. J. Role of
60. Kamal, S. M. et al. Specific cellular immune response and 85. Wilson, R. A., Coulson, P. S. & McHugh, S. M. A significant IL-6 in directing the initial immune response to schistosome
cytokine patterns in patients coinfected with hepatitis C virus part of the ‘concomitant immunity’ of mice to Schistosoma eggs. J. Immunol. 164, 2419–2426 (2000).
and Schistosoma mansoni. J. Infect. Dis. 184, 972–982 mansoni is the consequence of a leaky hepatic portal 107. Harris, D. P. et al. Reciprocal regulation of polarized cytokine
(2001). system, not immune killing. Parasite Immunol. 5, 595–601 production by effector B and T cells. Nature Immunol.
61. Kamal, S. M. et al. Acute hepatitis C without and with (1983). 1, 475–482 (2000).
schistosomiasis: correlation with hepatitis-C-specific CD4+ 86. Finkelman, F. D. & Urban, J. F. Jr. The other side of the coin: 108. Martin, D. L., King, C. L., Pearlman, E., Strine, E. & Heinzel,
T-cell and cytokine response. Gastroenterology 121, the protective role of the TH2 cytokines. J. Allergy Clin. F. P. IFN-γ is necessary, but not sufficient, for anti-CD40
646–656 (2001). Immunol. 107, 772–780 (2001). antibody-mediated inhibition of the TH2 response to
62. McClary, H., Koch, R., Chisari, F. V. & Guidotti, L. G. 87. Pearce, E. J., Casper, P., Grzych, J.-M., Lewis, F. A. & Sher, A. Schistosoma mansoni eggs. J. Immunol. 164, 779–785
Inhibition of hepatitis B virus replication during Schistosoma Downregulation of TH1 cytokine production accompanies (2000).
mansoni infection in transgenic mice. J. Exp. Med. 192, induction of TH2 responses by a parasitic helminth, 109. van Kooten, C. & Banchereau, J. Functions of CD40 on B
289–294 (2000). Schistosoma mansoni. J. Exp. Med. 173, 159–166 (1991). cells, dendritic cells and other cells. Curr. Opin. Immunol.
63. Brunet, L. R., Beall, M., Dunne, D. W. & Pearce, E. J. Nitric 88. Grzych, J. M. et al. Egg deposition is the major stimulus for 9, 330–337 (1997).
oxide and the TH2 response combine to prevent severe the production of TH2 cytokines in murine schistosomiasis 110. MacDonald, A. S., Straw, A. D., Dalton, N. M. & Pearce, E. J.
hepatic damage during Schistosoma mansoni infection. mansoni. J. Immunol. 146, 1322–1327 (1991). Cutting edge: TH2 response induction by dendritic cells: a
J. Immunol. 163, 4976–4984 (1999). References 87 and 88 were the first to show that role for CD40. J. Immunol. 168, 537–540 (2002).

510 | JULY 2002 | VOLUME 2 www.nature.com/reviews/immunol


REVIEWS

111. Coyle, A. J. & Gutierrez-Ramos, J. C. The expanding B7 dependent effector mechanisms. J. Immunol. 162, 345–351 143. Ishii, A. et al. Parasite infection and cancer: with special
superfamily: increasing complexity in costimulatory signals (1999). emphasis on Schistosoma japonicum infections
regulating T-cell function. Nature Immunol. 2, 203–209 128. Wynn, T. A. et al. IL-12 enhances vaccine-induced immunity (Trematoda). A review. Mutat. Res. 305, 273–281 (1994).
(2001). to schistosomes by augmenting both humoral and cell- 144. Feldmeier, H., Leutscher, P., Poggensee, G. & Harms, G.
112. Subramanian, G. et al. B7-2 requirement for helminth- mediated immune responses against the parasite. Male genital schistosomiasis and haemospermia. Trop.
induced granuloma formation and CD4 type-2 T helper cell J. Immunol. 157, 4068–4078 (1996). Med. Int. Health 4, 791–793 (1999).
cytokine expression. J. Immunol. 158, 5914–5920 (1997). 129. Chiaramonte, M. G., Hesse, M., Cheever, A. W. & Wynn, T. A. 145. Poggensee, G., Krantz, I., Kiwelu, I., Diedrich, T. &
113. Kopf, M. et al. Inducible costimulator protein (ICOS) controls CpG oligonucleotides can prophylactically immunize against Feldmeier, H. Presence of Schistosoma mansoni eggs in the
T-helper cell subset polarization after virus and parasite TH2-mediated schistosome egg-induced pathology by an cervix uteri of women in Mwanga District, Tanzania. Trans. R.
infection. J. Exp. Med. 192, 53–61 (2000). IL-12-independent mechanism. J. Immunol. 164, 973–985 Soc. Trop. Med. Hyg. 95, 299–300 (2001).
114. Xu, D. et al. Selective expression of a stable cell surface (2000). 146. Fallon, P. G. & Dunne, D. W. Tolerization of mice to
molecule on type 2 but not type 1 helper T cells. J. Exp. 130. Anderson, S., Shires, V. L., Wilson, R. A. & Mountford, A. P. Schistosoma mansoni egg antigens causes elevated type 1
Med. 187, 787–794 (1998). In the absence of IL-12, the induction of TH1-mediated and diminished type 2 cytokine responses and increased
115. Tesciuba, A. G. et al. Inducible costimulator regulates TH2- protective immunity by the attenuated schistosome vaccine mortality in acute infection. J. Immunol. 162, 4122–4132
mediated inflammation, but not TH2 differentiation, in a is impaired, revealing an alternative pathway with TH2-type (1999).
model of allergic airway disease. J. Immunol. 167, characteristics. Eur. J. Immunol. 28, 2827–2838 (1998). 147. Dunne, D. W. & Doenhoff, M. J. Schistosoma mansoni egg
1996–2003 (2001). 131. Mangold, B. L. & Dean, D. A. The role of IgG antibodies from antigens and hepatocyte damage in infected T-cell-deprived
116. Lohning, M. et al. T1/ST2 expression is enhanced on CD4+ irradiated cercaria-immunized rabbits in the passive transfer mice. Contrib. Microbiol. Immunol. 7, 22–29 (1983).
T cells from schistosome egg-induced granulomas: analysis of immunity to Schistosoma mansoni-infected mice. Am. J. 148. Read, S. & Powrie, F. CD4+ regulatory T cells. Curr. Opin.
of TH-cell cytokine coexpression ex vivo. J. Immunol. 162, Trop. Med. Hyg. 47, 821–829 (1992). Immunol. 13, 644–649 (2001).
3882–3889 (1999). 132. Hoffmann, K. F., James, S. L., Cheever, A. W. & Wynn, T. A. 149. Angeli, V. et al. Role of the parasite-derived prostaglandin D2
117. Townsend, M. J., Fallon, P. G., Matthews, D. J., Jolin, H. E. Studies with double cytokine-deficient mice reveal that in the inhibition of epidermal Langerhans cell migration
& McKenzie, A. N. T1/ST2-deficient mice demonstrate the highly polarized TH1- and TH2-type cytokine and antibody during schistosomiasis infection. J. Exp. Med. 193,
importance of T1/ST2 in developing primary T helper cell responses contribute equally to vaccine-induced immunity 1135–1147 (2001).
type-2 responses. J. Exp. Med. 191, 1069–1076 (2000). to Schistosoma mansoni. J. Immunol. 163, 927–938 (1999). 150. Fallon, P. G. Immunopathology of schistosomiasis: a
118. Capron, A., Capron, M., Dombrowicz, D. & Riveau, G. 133. Doenhoff, M., Kimani, G. & Cioli, D. Praziquantel and the cautionary tale of mice and men. Immunol. Today 21, 29–35
Vaccine strategies against schistosomiasis: from concepts control of schistosomiasis. Parasitol. Today 16, 364–366 (2000).
to clinical trials. Int. Arch. Allergy Immunol. 124, 9–15 (2001). (2000).
119. Bergquist, N. & Colley, D. Schistosomiasis vaccines: 134. Wynn, T. A. Development of an antipathology vaccine for Acknowledgements
research to development. Parasitol. Today 14, 99–104 schistosomiasis. Ann. NY Acad. Sci. 797, 191–195 (1996). E.J.P. is a Burroughs Wellcome Fund Scholar in Molecular
(1998). 135. Stadecker, M. J. The regulatory role of the antigen- Parasitology, and is supported by grants from the National Institutes
120. Pearce, E. Progress towards a vaccine for schistosomiasis. presenting cell in the development of hepatic of Health. A.S.M. is a recipient of a Wellcome Trust Prize Travelling
Acta Tropica (in the press). immunopathology during infection with Schistosoma Fellowship. We apologize to those authors whose work we were
121. Wilson, R. A., Coulson, P. S. & Mountford, A. P. Immune mansoni. Pathobiology 67, 269–272 (1999). unable to cite owing to space limitations.
responses to the radiation-attenuated schistosome vaccine: 136. Pearce, E. J. & Sher, A. Mechanisms of immune evasion in
what can we learn from knock-out mice? Immunol. Lett. 65, schistosomiasis. Contrib. Microbiol. Immunol. 8, 219–232
117–123 (1999). (1987). Online links
122. Wynn, T. A. & Hoffmann, K. F. Defining a schistosomiasis 137. Colley, D. G., LoVerde, P. T. & Savioli, L. Infectious disease.
vaccination strategy — is it really TH1 versus TH2? Parasitol. Medical helminthology in the 21st century. Science 293, DATABASES
Today 16, 497–501 (2000). 1437–1438 (2001). The following terms in this article are linked online to:
123. Caulada-Benedetti, Z., al-Zamel, F., Sher, A. & James, S. 138. Cribb, T. A., Bray, R. A., Littlewood, T., Pichelin, S. P. & Entrez: http://www.ncbi.nlm.nih.gov/Entrez/
Comparison of TH1- and TH2-associated immune reactivities Herniou, E. A. In Interrelationships of the Platyhelminthes HBV | HCV | HIV-1 | Mycobacterium bovis | Schistosoma
stimulated by single versus multiple vaccination of mice with (eds Littlewood, D. T. J. & Bray, R. A.) 168–185 (Taylor & japonicum | Schistosoma mansoni | Toxoplasma gondii | Trichuris
irradiated Schistosoma mansoni cercariae. J. Immunol. 146, Francis, London, 2001). muris |
1655–1660 (1991). 139. Agnew, A. M., Murare, H. M. & Doenhoff, M. J. Immune LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/
124. Oswald, I. P., Wynn, T. A., Sher, A. & James, S. L. NO as an attrition of adult schistosomes. Parasite Immunol. 15, B7RP1 | CD40 | CD80 | CD86 | CD154 | FcεR1 | ICOS | IFN-γ |
effector molecule of parasite killing: modulation of its 261–271 (1993). Ifnγ | IFN-γR1 | IL-1 | Il4 | Il4rα | IL-5 | IL-6 | IL-7 | Il7 | IL-10 | Il10 |
synthesis by cytokines. Comp. Biochem. Physiol. 140. Doenhoff, M. J. A role for granulomatous inflammation in the IL-12 | Il12 | IL-13 | Il13 | IL-13Rα2 | iNOS | MYD88 | OX40 |
Pharmacol. Toxicol. Endocrinol. 108, 11–18 (1994). transmission of infectious disease: schistosomiasis and OX40L | Rag | SM1 | SM2 | STAT6 | T1/ST2 | TGF-β | TLRs | TNF |
125. Street, M. et al. TNF is essential for the cell-mediated tuberculosis. Parasitology 115, S113–S125 (1997). TNFRI | TNFRII
protective immunity induced by the radiation-attenuated 141. Karanja, D. M., Colley, D. G., Nahlen, B. L., Ouma, J. H. &
schistosome vaccine. J. Immunol. 163, 4489–4494 (1999). Secor, W. E. Studies on schistosomiasis in western Kenya. I. FURTHER INFORMATION
126. Wilson, R. A., Coulson, P. S., Betts, C., Dowling, M. A. & Evidence for immune-facilitated excretion of schistosome The WHO — Schistosomiasis (Bilharzia):
Smythies, L. E. Impaired immunity and altered pulmonary eggs from patients with Schistosoma mansoni and human http://www.who.int/health-topics/schisto.htm
responses in mice with a disrupted interferon-γ receptor immunodeficiency virus coinfections. Am. J. Trop. Med. The WHO/UNDP/World Bank Schistosoma Genome
gene exposed to the irradiated Schistosoma mansoni Hyg. 56, 515–521 (1997). Network: http://www.nhm.ac.uk/hosted_sites/schisto/
vaccine. Immunology 87, 275–282 (1996). 142. Ngaiza, J. R. & Doenhoff, M. J. Blood platelets and University of Cambridge Schistosomiasis Research Group:
127. Jankovic, D. et al. Optimal vaccination against Schistosoma schistosome egg excretion. Proc. Soc. Exp. Biol. Med. 193, http://www.path.cam.ac.uk/~schisto/
mansoni requires the induction of both B-cell- and IFN-γ- 73–79 (1990). Access to this interactive links box is free online.

NATURE REVIEWS | IMMUNOLOGY VOLUME 2 | JULY 2002 | 5 1 1

View publication stats

You might also like